1
|
Hu Y, Hai J, Ti Y, Kong B, Yao G, Zhao Y, Zhang C, Zheng X, Zhang C, Ma X, Yu H, Qin X, Kovarik P, Zhang C, Liu S, Zhang W, Li J, Bu P. Adipose ZFP36 protects against diet-induced obesity and insulin resistance. Metabolism 2025; 164:156131. [PMID: 39761791 DOI: 10.1016/j.metabol.2024.156131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/21/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
AIMS Obesity, as a worldwide healthcare problem, has become more prevalent. ZFP36 is a well-known RNA-binding protein and involved in the posttranscriptional regulation of many physiological processes. Whether the adipose ZFP36 plays a role in obesity and insulin resistance remains unclear. METHODS The expression levels of ZFP36 were analyzed in adipose tissues of obese patients, diet-induced obese mice, ob/ob mice and db/db mice. To determine whether adipose ZFP36 protects against the diet-induced obesity, we generated adipose-specific ZFP36 knockout (ZFP36AKO) mice, which were subjected to high-fat-diet (HFD) for 16 weeks. To explore the specific molecular mechanisms of ZFP36 regulating metabolic disorders, we used gene array assay of control and ZFP36-deficient adipose tissue, and assessed the pathways in vitro and vivo. RESULTS Western blotting and RT-PCR were performed to determine the downregulating level of ZFP36 in adipose tissues of obese patients, diet-induced obese mice, ob/ob mice and db/db mice. Relative to control mice, ZFP36AKO mice were more susceptible to HFD-induced obesity, along with insulin resistance, glucose tolerance, and increased metabolic disorders. The obesity of ZFP36AKO mice was attributed to hypertrophy of adipocytes in white adipose tissue via decreased expression of Perilipin1 (PLIN1), adipose triglyceride lipase (ATGL), and hormone-sensitive lipase (HSL). We discovered that ZFP36 oppositely regulated RNF128 expression by repressing the mRNA stability and translation of RNF128, a negative regulator of Sirt1 expression. CONCLUSIONS This study suggests that ZFP36 in adipose tissue plays an important role in diet-induced obesity, and identifies a novel molecular signaling pathway of ZFP36/RNF128/Sirt1 involved in obesity.
Collapse
Affiliation(s)
- Yang Hu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jinghan Hai
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Binghui Kong
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Guoqing Yao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuan Zhao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Geriatric Medicine, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Xuehui Zheng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chunmei Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Xiangping Ma
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Huaitao Yu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoning Qin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Pavel Kovarik
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Cheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shaozhuang Liu
- Department of Bariatric and Metabolic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wencheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Jingyuan Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Peili Bu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
2
|
Wu Z, Wang Z, Chen T, Wang D, Zhou F, Zhang G, Wei S, Wu Y. Dermal white adipose tissue: A new modulator in wound healing and regeneration. Regen Ther 2025; 28:115-125. [PMID: 39717110 PMCID: PMC11665542 DOI: 10.1016/j.reth.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/25/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
Dermal white adipose tissue (dWAT), distinguished by its origin from cells within the dermis and independence from subcutaneous fat tissue, has garnered significant attention for its non-metabolic functions. Characterized by strong communication with other components of the skin, dWAT mediates the proliferation and recruitment of various cell types by releasing adipogenic and inflammatory factors. Here, we focus on the modulatory role of dWAT at different stages during wound healing, highlighting its ability to mediate the adipocyte-to-myofibroblast transition which plays a pivotal role in the physiology and pathology processes of skin fibrosis, scarring, and aging. This review highlights the regulatory potential of dWAT in modulating wound healing processes and presents it as a target for developing therapeutic strategies aimed at reducing scarring and enhancing regenerative outcomes in skin-related disorders.
Collapse
Affiliation(s)
- Zhongyu Wu
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Zhanqi Wang
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun Avenue South, Haidian District, Beijing 100081, PR China
| | - Tao Chen
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Dongyang Wang
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Feng Zhou
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Guorui Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Shan Wei
- Huizhou Health Sciences Polytechnic, Huizhou 516025, Guangdong, PR China
| | - Yingying Wu
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| |
Collapse
|
3
|
Huang CC, Wang CH, Yeh HY, Tsai HC, Yang CW, Li TH, Su CW, Yang YY, Lin HC, Hou MC. Peroxisome Proliferator-Activated Receptor α/γ and Cannabinoid Receptor 2 Agonist Attenuated Nonalcoholic Steatohepatitis Exosome-Related Abnormalities in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:188-203. [PMID: 39490440 DOI: 10.1016/j.ajpath.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/20/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
This study explored the mechanisms and effects of 1 month of peroxisome proliferator-activated receptor (PPAR)α/γ agonist aleglitazar (10 mg/kg per day) or cannabinoid receptor 2 (CB2R) agonist JWH015 (3 mg/kg per day), alone or combined, on visceral adipose tissue (VAT)-derived extracellular vesicle (EV) release and associated systemic/VAT inflammation, decreased VAT capillary density/fibrosis, and intestinal inflammation/hyperpermeability in nonalcoholic steatohepatitis (NASH) mice. High EV release from VAT of NASH mice was associated with severe systemic/VAT/intestinal inflammation, reduced capillary network of VAT, and intestinal hyperpermeability. Combined JWH015 with aleglitazar treatment suppressed high-fat diet-induced obesity/adiposity, inhibited VAT expansion, reduced VAT inflammation/fibrosis, normalized VAT capillary network, and attenuated intestinal mucosal injury, inflammation, and hyperpermeability in NASH + aleglitazar + JWH015 mice. The inhibition of adipose tissue (AT)-derived EV release and hypoxia-inducible factor (HIF)1α levels in AT-derived EV, normalization of CB2R, PPARα, PPARγ, PPARγ1, PPARγ2, tight junction proteins, vascular endothelial growth factor/CD31 expression, and down-regulation of HIF1α, monocyte chemoattractant protein-1, and transforming growth factor-β1 were observed in the VAT and intestine of the NASH + aleglitazar + jwh015 group. In vitro experiments revealed that PPARα/γ and CB2R activation attenuated NASH AT-derived EV-induced pathogenic changes in the J774/SVEC4-10/Caco2/3T3-L1 cell system. This study suggested that VAT-derived EVs contribute to the pathogenesis of NASH and that combined PPARα/γ and CB2R agonist treatment ameliorated the abovementioned abnormalities of NASH mice.
Collapse
Affiliation(s)
- Chia-Chang Huang
- Department of Medical Education, Taipei Veterans General Hospital, Taipei City, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan
| | - Ching-Hsiang Wang
- Department of Medical Education, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Hsiao-Yun Yeh
- Department of Medical Education, Taipei Veterans General Hospital, Taipei City, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan
| | - Hung-Cheng Tsai
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan; Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Ching-Wen Yang
- Department of Medical Education, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Tzu-Hao Li
- Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Foundation Hospital, Taipei City, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, Taipei City, Taiwan
| | - Chien-Wei Su
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Ying-Ying Yang
- Department of Medical Education, Taipei Veterans General Hospital, Taipei City, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan.
| | - Han-Chieh Lin
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Ming-Chih Hou
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan; Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| |
Collapse
|
4
|
Vieira FT, Cai Y, Gonzalez MC, Goodpaster BH, Prado CM, Haqq AM. Poor muscle quality: A hidden and detrimental health condition in obesity. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09941-0. [PMID: 39833502 DOI: 10.1007/s11154-025-09941-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Poor muscle quality (MQ) is a hidden health condition in obesity, commonly disregarded and underdiagnosed, associated with poor health-related outcomes. This narrative review provides an in-depth exploration of MQ in obesity, including definitions, available assessment methods and challenges, pathophysiology, association with health outcomes, and potential interventions. MQ is a broad term that can include imaging, histological, functional, or metabolic assessments, evaluating beyond muscle quantity. MQ assessment is highly heterogeneous and requires further standardization. Common definitions of MQ include 1) muscle-specific strength (or functional MQ), the ratio between muscle strength and muscle quantity, and 2) muscle composition (or morphological MQ), mainly evaluating muscle fat infiltration. An individual with obesity might still have normal or higher muscle quantity despite having poor MQ, and techniques for direct measurements are needed. However, the use of body composition and physical function assessments is still limited in clinical practice. Thus, more accessible techniques for assessing strength, muscle mass, and composition should be further explored. Obesity leads to adipocyte dysfunction, generating a low-grade chronic inflammatory state, which leads to mitochondrial dysfunction. Adipocyte and mitochondrial dysfunction result in metabolic dysfunction manifesting clinically as insulin resistance, dyslipidemia, and fat infiltration into organs such as muscle, which in excess is termed myosteatosis. Myosteatosis decreases muscle cell function and insulin sensitivity, creating a vicious cycle of inflammation and metabolic derangements. Myosteatosis increases the risk of poor muscle function, systemic metabolic complications, and mortality, presenting prognostic potential. Interventions shown to improve MQ include nutrition, physical activity/exercise, pharmacology, and metabolic and bariatric surgery.
Collapse
Affiliation(s)
- Flavio T Vieira
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Yuanjun Cai
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - M Cristina Gonzalez
- Postgraduate Program in Nutrition and Food, Federal University of Pelotas, Pelotas, Rio Grande Do Sul, Brazil
| | | | - Carla M Prado
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Andrea M Haqq
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
5
|
Han S, Huang J, Yang C, Feng J, Wang Y. The histone demethylase KDM6B links obstructive sleep apnea to idiopathic pulmonary fibrosis. FASEB J 2025; 39:e70306. [PMID: 39781582 PMCID: PMC11712539 DOI: 10.1096/fj.202402813r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025]
Abstract
Obstructive sleep apnea (OSA) is increasingly recognized for its link to idiopathic pulmonary fibrosis (IPF), though the underlying mechanisms remain poorly understood. Histone lysine demethylase 6B (KDM6B) may either prevent or promote organ fibrosis, but its specific role in IPF is yet to be clarified. This study aimed to investigate the function and mechanisms of KDM6B in IPF and the exacerbating effects of OSA. We assessed KDM6B levels in lung tissues from IPF patients, IPF mouse models, and a dual-hit model combining OSA-associated intermittent hypoxia (IH) with bleomycin (BLM) or TGF-β1. We evaluated pulmonary fibrosis, myofibroblast activation, and oxidative stress. KDM6B levels were elevated in lung tissues from IPF patients and BLM-treated mice, as well as in TGF-β1-stimulated myofibroblasts. Importantly, IH significantly worsened BLM-induced pulmonary fibrosis and TGF-β1-induced myofibroblast activation, further amplifying KDM6B expression both in vivo and in vitro. Inhibition of KDM6B reduced pulmonary fibrosis and decreased fibroblast activation and migration in IPF and dual-hit models. Mechanistically, KDM6B inhibition led to decreased NOX4 expression and reduced oxidative stress. KDM6B plays a critical role in promoting pulmonary fibrosis and mediating the exacerbating effects of OSA on this condition. Our findings identify KDM6B as a novel potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Shuangyu Han
- Department of Respiratory and Critical Care MedicineTianjin Medical University General HospitalTianjinChina
| | - Jie Huang
- Department of Respiratory and Critical Care MedicineTianjin Medical University General HospitalTianjinChina
| | - Changqing Yang
- Department of Respiratory and Critical Care MedicineTianjin Medical University General HospitalTianjinChina
| | - Jing Feng
- Department of Respiratory and Critical Care MedicineTianjin Medical University General HospitalTianjinChina
| | - Yubao Wang
- Department of Respiratory and Critical Care MedicineTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
6
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025:10.1038/s41574-024-01071-y. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
7
|
Wang L, Li J, Tang P, Zhu D, Tai L, Wang Y, Miyata T, Woodgett JR, Di LJ. GSK3β Deficiency Expands Obese Adipose Vasculature to Mitigate Metabolic Disorders. Circ Res 2025; 136:91-111. [PMID: 39629559 DOI: 10.1161/circresaha.124.325187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Maintaining a well-developed vascular system alongside adipose tissue (AT) expansion significantly reduces the risk of metabolic complications. Although GSK3β (glycogen synthase kinase-3 beta) is known for its role in various cellular processes, its specific functions in AT and regulation of body homeostasis have not been reported. METHODS GSK3β-floxed and GSK3α-floxed mice were crossed with adiponectin-Cre mice to generate GSK3β or GSK3α adipocyte-specific knockout mice (GSK3βADKO and GSK3αADKO). A comprehensive whole-body metabolism analysis was performed on obese GSK3βADKO mice induced by a high-fat diet. RNA sequencing was conducted on AT of both obese GSK3βADKO and GSK3αADKO mice. Various analyses, including vessel perfusion studies, lipolysis analysis, multiplex protein assays, in vitro protein phosphorylation assays, and whole-mount histology staining, were performed on AT of obese GSK3βADKO mice. Tube-formation experiments were performed using 3B-11 endothelial cells cultured in the conditional medium of matured adipocytes under hypoxic conditions. Chromatin precipitation and immunofluorescence studies were conducted using cultured adipocytes with GSK3 inhibition. RESULTS Our findings provide the first evidence that adipocyte-specific knockout of GSK3β expands AT vascularization and mitigates obesity-related metabolic disorders. GSK3β deficiency, but not GSK3α, in adipocytes activates AMPK (AMP-activated protein kinase), leading to increased phosphorylation and nuclear accumulation of HIF-2α, resulting in enhanced transcriptional regulation. Consequently, adipocytes increased VEGF (vascular endothelial growth factor) expression, which engages VEGFR2 on endothelial cells, promoting angiogenesis, expanding the vasculature, and improving vessel perfusion within obese AT. GSK3β deficiency promotes AT remodeling, shifting unhealthy adipocyte function toward a healthier state by increasing insulin-sensitizing hormone adiponectin and preserving healthy adipocyte function. These effects lead to reduced fibrosis, reactive oxygen species, and ER (endoplasmic reticulum) stress in obese AT and improve metabolic disorders associated with obesity. CONCLUSIONS Deletion of GSK3β in adipocytes activates the AMPK/HIF-2α/VEGF/VEGFR2 axis, promoting vasculature expansion within obese AT. This results in a significantly improved local microenvironment, reducing inflammation and effectively ameliorating metabolic disorders associated with obesity.
Collapse
Affiliation(s)
- Li Wang
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
- The Ministry of Education Frontiers Science Center for Precision Oncology (L.W., L.D.), University of Macau, China
- Proteomics, Metabolomics and Drug development core facility, Faculty of Health Sciences (L.W.), University of Macau, China
| | - Jiajia Li
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Ping Tang
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Dongliang Zhu
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Lixin Tai
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Yuan Wang
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
| | - Tsukiko Miyata
- Lunenfeld-Tanenbaum Research Institute, Sinai Health and Department of Medical Biophysics, University of Toronto, Ontario, Canada (T.M., J.R.W.)
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health and Department of Medical Biophysics, University of Toronto, Ontario, Canada (T.M., J.R.W.)
| | - Li-Jun Di
- Department of Biomedical Sciences, Faculty of Health Sciences (L.W., J.L., P.T., D.Z., L.T., Y.W., L.D.), University of Macau, China
- The Ministry of Education Frontiers Science Center for Precision Oncology (L.W., L.D.), University of Macau, China
| |
Collapse
|
8
|
Wang Z, Liu X, Sheng L, Xie Y, Feng W, Yu L. Effects of duration of high-fat diet on adipocyte hyperplasia in rat epididymis. Obes Res Clin Pract 2025; 19:54-62. [PMID: 39922761 DOI: 10.1016/j.orcp.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/21/2023] [Revised: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND High-fat diet (HFD) contributes to obesity and enhances the expression of mature adipocyte markers. However, the effect of HFD on adipocyte hyperplasia remains controversial. This may be due to variations in the duration of HFD. This study aimed to investigate the effects of different durations of HFD on adipocyte hyperplasia and the expression of mature adipocyte-related markers in obese rats. METHODS We divided 32 Sprague-Dawley rats into four groups: B (standard diet control), H1 (HFD for four weeks), H2 (HFD for eight weeks), and H3 (HFD for 12 weeks). We evaluated the morphological changes in epididymal fat cells, measured serum inflammatory markers using enzyme-linked immunosorbent assay (ELISA) kits, and quantified adipocyte hyperplasia and maturation markers using western blotting. RESULTS We observed progressive increases in body weight, epididymal fat weight, serum leptin, TNF-α, IL-6, irisin, PPARγ, adiponectin, and FNDC5 protein expression over 8 weeks of HFD. 12 weeks of HFD intervention resulted in significant decreases in irisin, PPARγ, adiponectin, and FNDC5. Concurrently, the expression of perilipin A and ATGL declined with prolonged HFD. CONCLUSIONS Our results suggest that the duration of HFD significantly affects adipocyte ability to undergo hyperplasia in the epididymis of obese rats. Specifically, 4 weeks of HFD did not change the capacity for adipocyte hyperplasia, while 8 weeks of the diet enhanced this capacity. Interestingly, a longer diet duration (12 weeks) led to a decrease in adipocyte hyperplasia.
Collapse
Affiliation(s)
- Zhaoxin Wang
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Xiujuan Liu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | - Lei Sheng
- Department of Scientific Research, Nanjing Sport Institute, Nanjing, China.
| | - Yuting Xie
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Wanyu Feng
- Department of Science Experiment Center, Nanjing Sport Institute, Nanjing, China
| | - Li Yu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| |
Collapse
|
9
|
Blüher M. Understanding Adipose Tissue Dysfunction. J Obes Metab Syndr 2024; 33:275-288. [PMID: 39734091 PMCID: PMC11704217 DOI: 10.7570/jomes24013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/25/2024] [Revised: 06/08/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
Diseases affecting adipose tissue (AT) function include obesity, lipodystrophy, and lipedema, among others. Both a lack of and excess AT are associated with increased risk for developing diseases including type 2 diabetes mellitus, hypertension, obstructive sleep apnea, and some types of cancer. However, individual risk of developing cardiometabolic and other 'obesity-related' diseases is not entirely determined by fat mass. Rather than excess fat accumulation, AT dysfunction may represent the mechanistic link between obesity and comorbid diseases. There are people who remain metabolically healthy despite obesity, whereas people with normal weight or very low subcutaneous AT mass may develop typically obesity-related diseases. AT dysfunction is characterized by adipocyte hypertrophy, impaired subcutaneous AT expandability (ectopic fat deposition), hypoxia, a variety of stress, inflammatory processes, and the release of proinflammatory, diabetogenic, and atherogenic signals. Genetic and environmental factors might contribute to AT heterogeneity either alone or via interaction with intrinsic biological factors. However, many questions remain regarding the mechanisms of AT dysfunction initiation and whether and how it could be reversed. Do AT signatures define clinically relevant subtypes of obesity? Is the cellular composition of AT associated with variation in obesity phenotypes? What roles do environmental compounds play in the manifestation of AT dysfunction? Answers to these and other questions may explain AT disease mechanisms and help to define strategies for improving AT health. This review focuses on recent advances in our understanding of AT biology.
Collapse
Affiliation(s)
- Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
10
|
Sheikh E, Liu Q, Burk D, Beavers WN, Fu X, Gartia MR. Mapping lipid species remodeling in high fat diet-fed mice: Unveiling adipose tissue dysfunction with Raman microspectroscopy. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159557. [PMID: 39128539 PMCID: PMC11380576 DOI: 10.1016/j.bbalip.2024.159557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/21/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Dysregulated lipid metabolism in obesity leads to adipose tissue expansion, a major contributor to metabolic dysfunction and chronic disease. Lipid metabolism and fatty acid changes play vital roles in the progression of obesity. In this proof-of-concept study, Raman techniques combined with histochemical imaging methods were utilized to analyze the impact of a high-fat diet (HFD) on different types of adipose tissue in mice, using a small sample size (n = 3 per group). After six weeks of high-fat diet (HFD) feeding, our findings showed hypertrophy, elevated collagen levels, and increased macrophage presence in the adipose tissues of the HFD group compared to the low-fat diet (LFD) group. Statistical analysis of Raman spectra revealed significantly lower unsaturated lipid levels and higher lipid to protein content in different fat pads (brown adipose tissue (BAT), subcutaneous white adipose tissue (SWAT), and visceral white adipose tissue (VWAT)) with HFD. Raman images of adipose tissues were analyzed using Empty modeling and DCLS methods to spatially profile unsaturated and saturated lipid species in the tissues. It revealed elevated levels of ω-3, ω-6, cholesterol, and triacylglycerols in BAT adipose tissues of HFD compared to LFD tissues. These findings indicated that while cholesterol, ω-6/ω-3 ratio, and triacylglycerol levels have risen in the SWAT and VWAT adipose tissues of the HFD group, the levels of ω-3 and ω-6 have decreased following the HFD. The study showed that Raman spectroscopy provided invaluable information at the molecular level for investigating lipid species remodeling and spatial mapping of adipose tissues during HFD.
Collapse
Affiliation(s)
- Elnaz Sheikh
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Qianglin Liu
- LSU AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - David Burk
- Cell Biology and Bioimaging Core, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - William N Beavers
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; Louisiana State University School of Veterinary Medicine, Mass Spectrometry Resource Center, Baton Rouge, LA 70803, USA
| | - Xing Fu
- LSU AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
11
|
Odeniyi IA, Ahmed B, Anbiah B, Hester G, Abraham PT, Lipke EA, Greene MW. An improved in vitro 3T3-L1 adipocyte model of inflammation and insulin resistance. Adipocyte 2024; 13:2414919. [PMID: 39415617 PMCID: PMC11487959 DOI: 10.1080/21623945.2024.2414919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/25/2023] [Revised: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Tumor necrosis factor alpha (TNF-α)/hypoxia-treated 3T3-L1 adipocytes have been used to model inflamed and insulin-resistant adipose tissue: this study examines gaps in the model. We tested whether modulating TNF-α/hypoxia treatment time could reduce cell death while still inducing inflammation and insulin resistance. Adipocytes were treated with TNF-α (12 h or 24 h) and incubated in a hypoxic chamber for 24 h. To examine maintenance of the phenotype over time, glucose and FBS were added at 24 h post initiation of treatment, and the cells were maintained for an additional 48 h. Untreated adipocytes were used as a control. Viability, insulin resistance, and inflammation were assessed using Live/Dead staining, RT-qPCR, ELISA, and glucose uptake assays. Treatment for 12 h with TNF-α in the presence of hypoxia resulted in an increase in the percentage of live cells compared to 24 h treated cells. Importantly, insulin resistance and inflammation were still induced in the 12 h treated adipocytes: the expression of the insulin sensitive and inflammatory genes was decreased and increased, respectively. In 72 h treated adipocytes, no significant differences were found in the viability, glucose uptake or insulin-sensitive and inflammatory gene expression. This study provides a modified approach to in vitro odeling adipocyte inflammation and insulin resistance. .
Collapse
Affiliation(s)
| | - Bulbul Ahmed
- Department of Nutritional Sciences, Auburn University, Auburn, AL, USA
| | - Benjamin Anbiah
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | - Grace Hester
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | - Peter T. Abraham
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | | | - Michael W. Greene
- Department of Nutritional Sciences, Auburn University, Auburn, AL, USA
| |
Collapse
|
12
|
Kang GS, Kim YE, Oh HR, Jo HJ, Bok S, Jeon YK, Cheon GJ, Roh TY, Chang YT, Park DJ, Ahn GO. Hypoxia-inducible factor-1α-deficient adipose-tissue macrophages produce the heat to mediate lipolysis of white adipose tissue through uncoupling protein-1. Lab Anim Res 2024; 40:37. [PMID: 39473019 PMCID: PMC11523771 DOI: 10.1186/s42826-024-00224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Uncoupling protein 1 (UCP1) is a proton uncoupler located across the mitochondrial membrane generally involved in thermogenesis of brown adipose tissues. Although UCP1 is known to be strongly expressed in brown adipocytes, recent evidence suggest that white adipocytes can also express UCP1 under certain circumstances such as cold- or β-adrenergic receptor-stimulation, allowing them to acquire brown adipocyte-like features thereby becoming 'beige' adipocytes. RESULTS In this study, we report that UCP1 can be expressed in adipose-tissue macrophages (ATM) lacking functional hypoxia-inducible factor-1 (HIF-1) and this does not require cold- nor β-adrenergic receptor activation. By using myeloid-specific Hif-1α knockout (KO) mice, we observed that these mice were protected from diet-induced obesity and exhibited an improved thermogenic tolerance upon cold challenge. ATM isolated from white adipose tissues (WAT) of these mice fed with high fat diet exhibited significantly higher M2-polarization, decreased glycolysis, increased mitochondrial functions and acetyl-CoA levels, along with increased expression of Ucp1, peroxisome proliferator activated receptor-gamma co-activator-1a, and others involved in histone acetylation. Consistent with the increased Ucp1 gene expression, these ATM produced a significant amount of heat mediating lipolysis of co-cultured adipocytes liberating free fatty acid. Treating ATM with acetate, a substrate for acetyl-CoA synthesis was able to boost the heat production in wild-type or Hif-1α-deficient but not UCP1-deficient macrophages, indicating that UCP1 was necessary for the heat production in macrophages. Lastly, we observed a significant inverse correlation between the number of UCP1-expressing ATM in WAT and the body mass index of human individuals. CONCLUSIONS UCP1-expressing ATM produce the heat to mediate lipolysis of adipocytes, indicating that this can be a novel strategy to treat and prevent diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Young-Eun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Ho Rim Oh
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Seoyeon Bok
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Tae-Young Roh
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Korea
| | | | - Do Joong Park
- Department of Surgery, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea.
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
13
|
Bahn YJ, Wang Y, Dagur P, Scott N, Cero C, Long KT, Nguyen N, Cypess AM, Rane SG. TGF-β antagonism synergizes with PPARγ agonism to reduce fibrosis and enhance beige adipogenesis. Mol Metab 2024; 90:102054. [PMID: 39461664 PMCID: PMC11570741 DOI: 10.1016/j.molmet.2024.102054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVES Adipose tissue depots vary markedly in their ability to store and metabolize triglycerides, undergo beige adipogenesis and susceptibility to metabolic disease. The molecular mechanisms that underlie such heterogeneity are not entirely clear. Previously, we showed that TGF-β signaling suppresses beige adipogenesis via repressing the recruitment of dedicated beige progenitors. Here, we find that TGF-β signals dynamically regulate the balance between adipose tissue fibrosis and beige adipogenesis. METHODS We investigated adipose tissue depot-specific differences in activation of TGF-β signaling in response to dietary challenge. RNA-seq and fluorescence activated cell sorting was performed to identify and characterize cells responding to changes in TGF-β signaling status. Mouse models, pharmacological strategies and human adipose tissue analyses were performed to further define the influence of TGF-β signaling on fibrosis and functional beige adipogenesis. RESULTS Elevated basal and high-fat diet inducible activation of TGF-β/Smad3 signaling was observed in the visceral adipose tissue depot. Activation of TGF-β/Smad3 signaling was associated with increased adipose tissue fibrosis. RNA-seq combined with fluorescence-activated cell sorting of stromal vascular fraction of epididymal white adipose tissue depot resulted in identification of TGF-β/Smad3 regulated ITGA5+ fibrogenic progenitors. TGF-β/Smad3 signal inhibition, genetically or pharmacologically, reduced fibrosis and increased functional beige adipogenesis. TGF-β/Smad3 antagonized the beneficial effects of PPARγ whereas TGF-β receptor 1 inhibition synergized with actions of rosiglitazone, a PPARγ agonist, to dampen fibrosis and promote beige adipogenesis. Positive correlation between TGF-β activation and ITGA5 was observed in human adipose tissue, with visceral adipose tissue depots exhibiting higher fibrosis potential than subcutaneous or brown adipose tissue depots. CONCLUSIONS Basal and high-fat diet inducible activation of TGF-β underlies the heterogeneity of adipose tissue depots. TGF-β/Smad3 activation promotes adipose tissue fibrosis and suppresses beige progenitors. Together, these dual mechanisms preclude functional beige adipogenesis. Controlled inhibition of TβRI signaling and concomitant PPARγ stimulation can suppress adipose tissue fibrosis and promote beige adipogenesis to improve metabolism.
Collapse
Affiliation(s)
- Young Jae Bahn
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Yanling Wang
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Pradeep Dagur
- Flow Cytometry Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Nicholas Scott
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Cheryl Cero
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Kelly T Long
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Nhuquynh Nguyen
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Sushil G Rane
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
14
|
Sinha SK, Carpio MB, Nicholas SB. Fiery Connections: Macrophage-Mediated Inflammation, the Journey from Obesity to Type 2 Diabetes Mellitus and Diabetic Kidney Disease. Biomedicines 2024; 12:2209. [PMID: 39457523 PMCID: PMC11503991 DOI: 10.3390/biomedicines12102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
The high prevalence of diabetes mellitus (DM) poses a significant public health challenge, with diabetic kidney disease (DKD) as one of its most serious consequences. It has become increasingly clear that type 2 DM (T2D) and the complications of DKD are not purely metabolic disorders. This review outlines emerging evidence related to the step-by-step contribution of macrophages to the development and progression of DKD in individuals who specifically develop T2D as a result of obesity. The macrophage is a prominent inflammatory cell that contributes to obesity, where adipocyte hypertrophy leads to macrophage recruitment and eventually to the expansion of adipose tissue. The recruited macrophages secrete proinflammatory cytokines, which cause systemic inflammation, glucose dysregulation, and insulin sensitivity, ultimately contributing to the development of T2D. Under such pathological changes, the kidney is susceptible to elevated glucose and thereby activates signaling pathways that ultimately drive monocyte recruitment. In particular, the early recruitment of proinflammatory macrophages in the diabetic kidney produces inflammatory cytokines/chemokines that contribute to inflammation and tissue damage associated with DKD pathology. Macrophage activation and recruitment are crucial inciting factors that also persist as DKD progresses. Thus, targeting macrophage activation and function could be a promising therapeutic approach, potentially offering significant benefits for managing DKD at all stages of progression.
Collapse
Affiliation(s)
- Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
- Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Maria Beatriz Carpio
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Susanne B. Nicholas
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
15
|
Mo YY, Han YX, Xu SN, Jiang HL, Wu HX, Cai JM, Li L, Bu YH, Xiao F, Liang HD, Wen Y, Liu YZ, Yin YL, Zhou HD. Adipose Tissue Plasticity: A Comprehensive Definition and Multidimensional Insight. Biomolecules 2024; 14:1223. [PMID: 39456156 PMCID: PMC11505740 DOI: 10.3390/biom14101223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Adipose tissue is composed of adipocytes, stromal vascular fraction, nerves, surrounding immune cells, and the extracellular matrix. Under various physiological or pathological conditions, adipose tissue shifts cellular composition, lipid storage, and organelle dynamics to respond to the stress; this remodeling is called "adipose tissue plasticity". Adipose tissue plasticity includes changes in the size, species, number, lipid storage capacity, and differentiation function of adipocytes, as well as alterations in the distribution and cellular composition of adipose tissue. This plasticity has a major role in growth, obesity, organismal protection, and internal environmental homeostasis. Moreover, certain thresholds exist for this plasticity with significant individualized differences. Here, we comprehensively elaborate on the specific connotation of adipose tissue plasticity and the relationship between this plasticity and the development of many diseases. Meanwhile, we summarize possible strategies for treating obesity in response to adipose tissue plasticity, intending to provide new insights into the dynamic changes in adipose tissue and contribute new ideas to relevant clinical problems.
Collapse
Affiliation(s)
- Yu-Yao Mo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Xin Han
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Shi-Na Xu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Jun-Min Cai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yan-Hong Bu
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha 410012, China;
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Han-Dan Liang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Ying Wen
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Ze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
| | - Yu-Long Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| |
Collapse
|
16
|
Ahn C, Zhang T, Yang G, Rode T, Varshney P, Ghayur SJ, Chugh OK, Jiang H, Horowitz JF. Years of endurance exercise training remodel abdominal subcutaneous adipose tissue in adults with overweight or obesity. Nat Metab 2024; 6:1819-1836. [PMID: 39256590 DOI: 10.1038/s42255-024-01103-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/09/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024]
Abstract
Abnormalities in the structure and metabolic function of abdominal subcutaneous adipose tissue (aSAT) underlie many obesity-related health complications. Endurance exercise improves cardiometabolic health in adults with overweight or obesity, but the effects of endurance training on aSAT are unclear. We included male and female participants who were regular exercisers with overweight or obesity who exercised for >2 years, and cross-sectionally compared them with well-matched non-exercisers with overweight or obesity. Here we show aSAT from exercisers has a higher capillary density, lower Col6a abundance and fewer macrophages compared with non-exercisers. This is accompanied by a greater abundance of angiogenic, ribosomal, mitochondrial and lipogenic proteins. The abundance of phosphoproteins involved in protein translation, lipogenesis and direct regulation of transcripts is also greater in aSAT collected from exercisers. Exploratory ex vivo experiments demonstrate greater angiogenic capacity and higher lipid-storage capacity in samples cultured from aSAT collected from exercisers versus non-exercisers. Regular exercise may play a role in remodelling aSAT structure and proteomic profile in ways that may contribute to preserved cardiometabolic health.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gayoung Yang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Rode
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Pallavi Varshney
- Human Bioenergetics Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Sophia J Ghayur
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Wang R, Gomez Salazar M, Pruñonosa Cervera I, Coutts A, French K, Pinto MM, Gohlke S, García-Martín R, Blüher M, Schofield CJ, Kourtzelis I, Stimson RH, Bénézech C, Christian M, Schulz TJ, Gudmundsson EF, Jennings LL, Gudnason VG, Chavakis T, Morton NM, Emilsson V, Michailidou Z. Adipocyte deletion of the oxygen-sensor PHD2 sustains elevated energy expenditure at thermoneutrality. Nat Commun 2024; 15:7483. [PMID: 39209825 PMCID: PMC11362468 DOI: 10.1038/s41467-024-51718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/09/2022] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Enhancing thermogenic brown adipose tissue (BAT) function is a promising therapeutic strategy for metabolic disease. However, predominantly thermoneutral modern human living conditions deactivate BAT. We demonstrate that selective adipocyte deficiency of the oxygen-sensor HIF-prolyl hydroxylase (PHD2) gene overcomes BAT dormancy at thermoneutrality. Adipocyte-PHD2-deficient mice maintain higher energy expenditure having greater BAT thermogenic capacity. In human and murine adipocytes, a PHD inhibitor increases Ucp1 levels. In murine brown adipocytes, antagonising the major PHD2 target, hypoxia-inducible factor-(HIF)-2a abolishes Ucp1 that cannot be rescued by PHD inhibition. Mechanistically, PHD2 deficiency leads to HIF2 stabilisation and binding of HIF2 to the Ucp1 promoter, thus enhancing its expression in brown adipocytes. Serum proteomics analysis of 5457 participants in the deeply phenotyped Age, Gene and Environment Study reveal that serum PHD2 associates with increased risk of metabolic disease. Here we show that adipose-PHD2-inhibition is a therapeutic strategy for metabolic disease and identify serum PHD2 as a disease biomarker.
Collapse
Affiliation(s)
- Rongling Wang
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mario Gomez Salazar
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Iris Pruñonosa Cervera
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amanda Coutts
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Karen French
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Marlene Magalhaes Pinto
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Ruben García-Martín
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC (CNB-CSIC), Campus-UAM, Madrid, Spain
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research University of Oxford, Oxford, UK
| | - Ioannis Kourtzelis
- Hull York Medical School, York Biomedical Research Institute, University of York, York, UK
| | - Roland H Stimson
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Christian
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | | | - Lori L Jennings
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Vilmundur G Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Triantafyllos Chavakis
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Nicholas M Morton
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Zoi Michailidou
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK.
| |
Collapse
|
18
|
Wu L, Huang F, Yang L, Yang L, Sun Z, Zhang J, Xia S, Zhao H, Ding Y, Bian D, Li K. Interplay of FXN expression and lipolysis in white adipocytes plays a critical role in insulin sensitivity in Friedreich's ataxia mouse model. Sci Rep 2024; 14:19876. [PMID: 39191875 DOI: 10.1038/s41598-024-71099-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
Frataxin (FXN) is required for iron-sulfur cluster biogenesis, and its loss causes the early-onset neurodegenerative disease Friedreich ataxia (FRDA). Loss of FXN is a susceptibility factor in the development of diabetes, a common metabolic complication after myocardial hypertrophy in patients with FRDA. The underlying mechanism of FXN deficient-induced hyperglycemia in FRDA is, however, poorly understood. In this study, we confirmed that the FXN deficiency mouse model YG8R develops insulin resistance in elder individuals by disturbing lipid metabolic homeostasis in adipose tissues. Evaluation of lipolysis, lipogenesis, and fatty acid β-oxidation showed that lipolysis is most severely affected in white adipose tissues. Consistently, FXN deficiency significantly decreased expression of lipolytic genes encoding adipose triglyceride lipase (Atgl) and hormone-sensitive lipase (Hsl) resulting in adipocyte enlargement and inflammation. Lipolysis induction by fasting or cold exposure remarkably upregulated FXN expression, though FXN deficiency lessened the competency of lipolysis compared with the control or wild type mice. Moreover, we found that the impairment of lipolysis was present at a young age, a few months earlier than hyperglycemia and insulin resistance. Forskolin, an activator of lipolysis, or pioglitazone, an agonist of PPARγ, improved insulin sensitivity in FXN-deficient adipocytes or mice. We uncovered the interplay between FXN expression and lipolysis and found that impairment of lipolysis, particularly the white adipocytes, is an early event, likely, as a primary cause for insulin resistance in FRDA patients at later age.
Collapse
Affiliation(s)
- Lin Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Fei Huang
- Endocrinology Department, Yancheng First People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, People's Republic of China
| | - Lu Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Liu Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Zichen Sun
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Jinghua Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Siyu Xia
- Endocrinology Department, Yancheng First People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, People's Republic of China
| | - Hongting Zhao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Yibing Ding
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Dezhi Bian
- Endocrinology Department, Yancheng First People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, People's Republic of China.
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
19
|
Greco F, Panunzio A, Bernetti C, Tafuri A, Beomonte Zobel B, Mallio CA. The Radiogenomic Landscape of Clear Cell Renal Cell Carcinoma: Insights into Lipid Metabolism through Evaluation of ADFP Expression. Diagnostics (Basel) 2024; 14:1667. [PMID: 39125543 PMCID: PMC11311402 DOI: 10.3390/diagnostics14151667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
This study aims to explore the relationship between radiological imaging and genomic characteristics in clear cell renal cell carcinoma (ccRCC), focusing on the expression of adipose differentiation-related protein (ADFP) detected through computed tomography (CT). The goal is to establish a radiogenomic lipid profile and understand its association with tumor characteristics. Data from The Cancer Genome Atlas (TCGA) and the Cancer Imaging Archive (TCIA) were utilized to correlate imaging features with adipose differentiation-related protein (ADFP) expression in ccRCC. CT scans assessed various tumor features, including size, composition, margin, necrosis, and growth pattern, alongside measurements of tumoral Hounsfield units (HU) and abdominal adipose tissue compartments. Statistical analyses compared demographics, clinical-pathological features, adipose tissue quantification, and tumoral HU between groups. Among 197 patients, 22.8% exhibited ADFP expression significantly associated with hydronephrosis. Low-grade ccRCC patients expressing ADFP had higher quantities of visceral and subcutaneous adipose tissue and lower tumoral HU values compared to their high-grade counterparts. Similar trends were observed in low-grade ccRCC patients without ADFP expression. ADFP expression in ccRCC correlates with specific imaging features such as hydronephrosis and altered adipose tissue distribution. Low-grade ccRCC patients with ADFP expression display a distinct lipid metabolic profile, emphasizing the relationship between radiological features, genomic expression, and tumor metabolism. These findings suggest potential for personalized diagnostic and therapeutic strategies targeting tumor lipid metabolism.
Collapse
Affiliation(s)
- Federico Greco
- Department of Radiology, Cittadella della Salute, Azienda Sanitaria Locale di Lecce, Piazza Filippo Bottazzi, 2, 73100 Lecce, Italy
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (C.B.); (B.B.Z.); (C.A.M.)
| | - Andrea Panunzio
- Department of Urology, “Vito Fazzi” Hospital, Piazza Filippo Muratore, 1, 73100 Lecce, Italy; (A.P.); (A.T.)
| | - Caterina Bernetti
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Alessandro Tafuri
- Department of Urology, “Vito Fazzi” Hospital, Piazza Filippo Muratore, 1, 73100 Lecce, Italy; (A.P.); (A.T.)
| | - Bruno Beomonte Zobel
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Carlo Augusto Mallio
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| |
Collapse
|
20
|
Mashayekhi M, Sheng Q, Bailin SS, Massier L, Zhong J, Shi M, Wanjalla CN, Wang TJ, Ikizler TA, Niswender KD, Gabriel CL, Palacios J, Turgeon-Jones R, Reynolds CF, Luther JM, Brown NJ, Das S, Dahlman I, Mosley JD, Koethe JR, Rydén M, Bachmann KN, Shah RV. The subcutaneous adipose transcriptome identifies a molecular signature of insulin resistance shared with visceral adipose. Obesity (Silver Spring) 2024; 32:1526-1540. [PMID: 38967296 PMCID: PMC11269023 DOI: 10.1002/oby.24064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/09/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE The objective of this study was to identify the transcriptional landscape of insulin resistance (IR) in subcutaneous adipose tissue (SAT) in humans across the spectrum of obesity. METHODS We used SAT RNA sequencing in 220 individuals with metabolic phenotyping. RESULTS We identified a 35-gene signature with high predictive accuracy for homeostatic model of IR that was expressed across a variety of non-immune cell populations. We observed primarily "protective" IR associations for adipocyte transcripts and "deleterious" associations for macrophage transcripts, as well as a high concordance between SAT and visceral adipose tissue (VAT). Multiple SAT genes exhibited dynamic expression 5 years after weight loss surgery and with insulin stimulation. Using available expression quantitative trait loci in SAT and/or VAT, we demonstrated similar genetic effect sizes of SAT and VAT on type 2 diabetes and BMI. CONCLUSIONS SAT is conventionally viewed as a metabolic buffer for lipid deposition during positive energy balance, whereas VAT is viewed as a dominant contributor to and prime mediator of IR and cardiometabolic disease risk. Our results implicate a dynamic transcriptional architecture of IR that resides in both immune and non-immune populations in SAT and is shared with VAT, nuancing the current VAT-centric concept of IR in humans.
Collapse
Affiliation(s)
- Mona Mashayekhi
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
| | - Quanhu Sheng
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, Tennessee, USA
| | - Samuel S. Bailin
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, USA
| | - Lucas Massier
- Karolinska Institutet, Department of Medicine, Huddinge (H7), Stockholm, Sweden
| | - Jiawei Zhong
- Karolinska Institutet, Department of Medicine, Huddinge (H7), Stockholm, Sweden
| | - Mingjian Shi
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Celestine N. Wanjalla
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, USA
| | - Thomas J. Wang
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, USA
| | - T. Alp Ikizler
- Vanderbilt University Medical Center, Department of Medicine, Division of Nephrology and Hypertension, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Kevin D. Niswender
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Curtis L. Gabriel
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Nashville, Tennessee, USA
| | - Julia Palacios
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
| | - Rachel Turgeon-Jones
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
| | - Cassandra F. Reynolds
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Department of Medicine, Cardiology Division, Nashville, Tennessee, USA
| | - James M. Luther
- Vanderbilt University Medical Center, Department of Medicine, Division of Nephrology and Hypertension, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Saumya Das
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Ingrid Dahlman
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan D. Mosley
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John R. Koethe
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine, Huddinge (H7), Stockholm, Sweden
| | - Katherine N. Bachmann
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Ravi V. Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Department of Medicine, Cardiology Division, Nashville, Tennessee, USA
| |
Collapse
|
21
|
Subošić B, Zdravković V, Ješić M, Munjas J, Kovačević S, Guzonjić A, Mitrović J, Saso L, Đuričić I, Kotur-Stevuljević J. Childhood obesity accelerates biological ageing: is oxidative stress a link? Br J Nutr 2024; 132:227-235. [PMID: 38736405 DOI: 10.1017/s0007114524000898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/14/2024]
Abstract
Obesity is a multifactorial pathophysiological condition with an imbalance in biochemical, immunochemical, redox status and genetic parameters values. We aimed to estimate the connection between relative leucocyte telomere lengths (rLTL) - biomarker of cellular ageing with metabolic and redox status biomarkers values in a group of obese and lean children. The study includes 110 obese and 42 lean children and adolescents, both sexes. The results suggested that rLTL are significantly shorter in obese, compared with lean group (P < 0·01). Negative correlation of rLTL with total oxidant status (TOS) (Spearman's ρ = -0·365, P < 0·001) as well as with C-reactive protein (Spearman's ρ = -0·363, P < 0·001) were observed. Principal component analysis (PCA) extracted three distinct factors (i.e. principal components) entitled as: prooxidant factor with 35 % of total variability; antioxidant factor with 30 % of total variability and lipid antioxidant - biological ageing factor with 12 % of the total variability. The most important predictor of BMI > 30 kg/m2 according to logistic regression analysis was PCA-derived antioxidant factor's score (OR: 1·66, 95th Cl 1·05-2·6, P = 0·029). PCA analysis confirmed that oxidative stress importance in biological ageing is caused by obesity and its multiple consequences related to prooxidants augmentation and antioxidants exhaustion and gave us clear signs of disturbed cellular homoeostasis deepness, even before any overt disease occurrence.
Collapse
Affiliation(s)
- Branko Subošić
- Biochemical Laboratory, University Children's Hospital, Tiršova 10, Belgrade, Serbia
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000, Belgrade
| | - Vera Zdravković
- Department of Endocrinology, University Children's Hospital, Belgrade School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Department of Endocrinology, University Children's Hospital, Belgrade, 11000, Serbia
| | - Maja Ješić
- Department of Endocrinology, University Children's Hospital, Belgrade School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Department of Endocrinology, University Children's Hospital, Belgrade, 11000, Serbia
| | - Jelena Munjas
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000, Belgrade
| | - Smiljka Kovačević
- Department of Endocrinology, University Children's Hospital, Belgrade, 11000, Serbia
| | - Azra Guzonjić
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000, Belgrade
| | - Jadranka Mitrović
- Biochemical Laboratory, University Children's Hospital, Tiršova 10, Belgrade, Serbia
| | - Luciano Saso
- Department of Physiology and Pharmacology 'Vittorio Erspamer', Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185Rome, Italy
| | - Ivana Đuričić
- Department of Bromatology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000, Belgrade, Serbia
| | - Jelena Kotur-Stevuljević
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000, Belgrade
| |
Collapse
|
22
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
23
|
Hu J, Zheng S, Hua M, Ding M, Hu Z, Jiang H. Deletion of Impdh2 in adipocyte precursors limits the expansion of white adipose tissue and enhances metabolic health with overnutrition. Biochem Biophys Res Commun 2024; 716:149998. [PMID: 38692012 DOI: 10.1016/j.bbrc.2024.149998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
The equilibrium between the hypertrophic growth of existing adipocytes and adipogenesis is vital in managing metabolic stability in white adipocytes when faced with overnutrition. Adipogenesis has been established as a key player in combating metabolic irregularities caused by various factors. However, the benefits of increasing adipogenesis-mediated white adipose tissue (WAT) expansion for metabolic health regulation remain uncertain. Our findings reveal an increase in Impdh2 expression during the adipogenesis phase, both in vivo and in vitro. Xmp enhances adipogenic potential by fostering mitotic clonal expansion (MCE). The conditional knockout of Impdh2 in adipocyte progenitor cells(APCs) in adult and aged mice effectively curbs white adipose tissue expansion, ameliorates glucose tolerance, and augments energy expenditure under high-fat diet (HFD). However, no significant difference is observed under normal chow diet (NCD). Concurrently, the knockout of Impdh2 in APCs significantly reduces the count of new adipocytes induced by HFD, without affecting adipocyte size. Mechanistically, Impdh2 regulates the proliferation of APCs during the MCE phase via Xmp. Exogenous Xmp can significantly offset the reduction in adipogenic abilities of APCs due to Impdh2 deficiency. In summary, we discovered that adipogenesis-mediated WAT expansion, induced by overnutrition, also contributes to metabolic abnormalities. Moreover, the pivotal role of Impdh2 in regulating adipogenesis in APCs offers a novel therapeutic approach to combat obesity.
Collapse
Affiliation(s)
- Jiaqi Hu
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Shiqiong Zheng
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Mengting Hua
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Mei Ding
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China
| | - Zhangfeng Hu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Bio-medical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China; Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, 430056, Hubei, China.
| | - Haochen Jiang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Bio-medical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, China.
| |
Collapse
|
24
|
Gianazza E, Brioschi M, Eligini S, Banfi C. Mass spectrometry for the study of adipocyte cell secretome in cardiovascular diseases. MASS SPECTROMETRY REVIEWS 2024; 43:752-781. [PMID: 36161723 DOI: 10.1002/mas.21812] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/27/2022] [Revised: 08/04/2022] [Accepted: 09/03/2022] [Indexed: 06/16/2023]
Abstract
Adipose tissue is classically considered the primary site of lipid storage, but in recent years has garnered appreciation for its broad role as an endocrine organ, capable of remotely signaling to other tissues to alter their metabolic program. The adipose tissue is now recognized as a crucial regulator of cardiovascular health, mediated by the secretion of several bioactive products, with a wide range of endocrine and paracrine effects on the cardiovascular system. Thanks to the development and improvement of high-throughput mass spectrometry, the size and components of the human secretome have been characterized. In this review, we summarized the recent advances in mass spectrometry-based studies of the cell and tissue secretome for the understanding of adipose tissue biology, which may help to decipher the complex molecular mechanisms controlling the crosstalk between the adipose tissue and the cardiovascular system, and their possible clinical translation.
Collapse
Affiliation(s)
- Erica Gianazza
- Centro Cardiologico Monzino IRCCS, Unit of Functional Proteomics, Metabolomics and Network Analysis, Milan, Italy
| | - Maura Brioschi
- Centro Cardiologico Monzino IRCCS, Unit of Functional Proteomics, Metabolomics and Network Analysis, Milan, Italy
| | - Sonia Eligini
- Centro Cardiologico Monzino IRCCS, Unit of Functional Proteomics, Metabolomics and Network Analysis, Milan, Italy
| | - Cristina Banfi
- Centro Cardiologico Monzino IRCCS, Unit of Functional Proteomics, Metabolomics and Network Analysis, Milan, Italy
| |
Collapse
|
25
|
Pilger BI, Castro A, Vasconcellos FF, Moura KF, Signini ÉDF, Marqueze LFB, Fiorenza-Neto EA, Rocha MT, Pedroso GS, Cavaglieri CR, Ferreira AG, Figueiredo C, Minuzzi LG, Gatti da Silva GH, Castro GS, Lira FS, Seelaender M, Pinho RA. Obesity-dependent molecular alterations in fatal COVID-19: A retrospective postmortem study of metabolomic profile of adipose tissue. J Cell Biochem 2024; 125:e30566. [PMID: 38591648 DOI: 10.1002/jcb.30566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
We investigated the effects of obesity on metabolic, inflammatory, and oxidative stress parameters in the adipose tissue of patients with fatal COVID-19. Postmortem biopsies of subcutaneous adipose tissue were obtained from 25 unvaccinated inpatients who passed from COVID-19, stratified as nonobese (N-OB; body mass index [BMI], 26.5 ± 2.3 kg m-2) or obese (OB BMI 34.2 ± 5.1 kg m-2). Univariate and multivariate analyses revealed that body composition was responsible for most of the variations detected in the metabolome, with greater dispersion observed in the OB group. Fifteen metabolites were major segregation factors. Results from the OB group showed higher levels of creatinine, myo-inositol, O-acetylcholine, and succinate, and lower levels of sarcosine. The N-OB group showed lower levels of glutathione peroxidase activity, as well as higher content of IL-6 and adiponectin. We revealed significant changes in the metabolomic profile of the adipose tissue in fatal COVID-19 cases, with high adiposity playing a key role in these observed variations. These findings highlight the potential involvement of metabolic and inflammatory pathways, possibly dependent on hypoxia, shedding light on the impact of obesity on disease pathogenesis and suggesting avenues for further research and possible therapeutic targets.
Collapse
Affiliation(s)
- Bruna I Pilger
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Alex Castro
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
- Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Franciane F Vasconcellos
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Karen F Moura
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Étore De Favari Signini
- Cardiovascular Physical Therapy Laboratory, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Luis Felipe B Marqueze
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Edson A Fiorenza-Neto
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Mateus T Rocha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Giulia S Pedroso
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Claudia R Cavaglieri
- Exercise Physiology Laboratory, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | - Antonio G Ferreira
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Caique Figueiredo
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Luciele G Minuzzi
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Guilherme H Gatti da Silva
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Gabriela S Castro
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Fábio S Lira
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Ricardo A Pinho
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
26
|
Tan J, Virtue S, Norris DM, Conway OJ, Yang M, Bidault G, Gribben C, Lugtu F, Kamzolas I, Krycer JR, Mills RJ, Liang L, Pereira C, Dale M, Shun-Shion AS, Baird HJ, Horscroft JA, Sowton AP, Ma M, Carobbio S, Petsalaki E, Murray AJ, Gershlick DC, Nathan JA, Hudson JE, Vallier L, Fisher-Wellman KH, Frezza C, Vidal-Puig A, Fazakerley DJ. Limited oxygen in standard cell culture alters metabolism and function of differentiated cells. EMBO J 2024; 43:2127-2165. [PMID: 38580776 PMCID: PMC11148168 DOI: 10.1038/s44318-024-00084-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2024] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 04/07/2024] Open
Abstract
The in vitro oxygen microenvironment profoundly affects the capacity of cell cultures to model physiological and pathophysiological states. Cell culture is often considered to be hyperoxic, but pericellular oxygen levels, which are affected by oxygen diffusivity and consumption, are rarely reported. Here, we provide evidence that several cell types in culture actually experience local hypoxia, with important implications for cell metabolism and function. We focused initially on adipocytes, as adipose tissue hypoxia is frequently observed in obesity and precedes diminished adipocyte function. Under standard conditions, cultured adipocytes are highly glycolytic and exhibit a transcriptional profile indicative of physiological hypoxia. Increasing pericellular oxygen diverted glucose flux toward mitochondria, lowered HIF1α activity, and resulted in widespread transcriptional rewiring. Functionally, adipocytes increased adipokine secretion and sensitivity to insulin and lipolytic stimuli, recapitulating a healthier adipocyte model. The functional benefits of increasing pericellular oxygen were also observed in macrophages, hPSC-derived hepatocytes and cardiac organoids. Our findings demonstrate that oxygen is limiting in many terminally-differentiated cell types, and that considering pericellular oxygen improves the quality, reproducibility and translatability of culture models.
Collapse
Affiliation(s)
- Joycelyn Tan
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Sam Virtue
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Dougall M Norris
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Olivia J Conway
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ming Yang
- MRC Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, 50931, Germany
| | - Guillaume Bidault
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Christopher Gribben
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Fatima Lugtu
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Ioannis Kamzolas
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - James R Krycer
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| | - Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| | - Lu Liang
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Conceição Pereira
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Martin Dale
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Amber S Shun-Shion
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Harry Jm Baird
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - James A Horscroft
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - Marcella Ma
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Stefania Carobbio
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- Centro de Investigacion Principe Felipe, Valencia, 46012, Spain
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - David C Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Kelsey H Fisher-Wellman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, 27834, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, 50931, Germany
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Centro de Investigacion Principe Felipe, Valencia, 46012, Spain.
| | - Daniel J Fazakerley
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
27
|
Han Y, Ji B, Leng Y, Xie C. Inhibited hypoxia-inducible factor by intraoperative hyperglycemia increased postoperative delirium of aged patients: A review. Medicine (Baltimore) 2024; 103:e38349. [PMID: 39259057 PMCID: PMC11142828 DOI: 10.1097/md.0000000000038349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/21/2023] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 09/12/2024] Open
Abstract
The underlying mechanism of postoperative delirium (POD) in elderly people remains unclear. Perioperative hyperglycemia (POHG) is an independent risk indicator for POD, particularly in the elderly. Under cerebral desaturation (hypoxia) during general anesthesia, hypoxia-inducible factor (HIF) is neuroprotective during cerebral hypoxia via diverse pathways, like glucose metabolism and angiogenesis. Hyperglycemia can repress HIF expression and activity. On the other hand, POHG occurred among patients undergoing surgery. For surgical stress, hypothalamic-pituitary-adrenal activation and sympathoadrenal activation may increase endogenous glucose production via gluconeogenesis and glycogenolysis. Thus, under the setting of cerebral hypoxia during general anesthesia, we speculate that POHG prevents HIF-1α levels and function in the brain of aged patients, thus exacerbating the hypoxic response of HIF-1 and potentially contributing to POD. This paper sketches the underlying mechanisms of HIF in POD in elderly patients and offers novel insights into targets for preventing or treating POD in the same way as POHG.
Collapse
Affiliation(s)
- Yutong Han
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Bing Ji
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Pain Management, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Yulin Leng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
28
|
Priscilla L, Yoo C, Jang S, Park S, Lim G, Kim T, Lee DY. Immunotherapy targeting the obese white adipose tissue microenvironment: Focus on non-communicable diseases. Bioact Mater 2024; 35:461-476. [PMID: 38404641 PMCID: PMC10884763 DOI: 10.1016/j.bioactmat.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/29/2023] [Revised: 01/14/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Obesity triggers inflammatory responses in the microenvironment of white adipose tissue, resulting in chronic systemic inflammation and the subsequent development of non-communicable diseases, including type 2 diabetes, coronary heart disease, and breast cancer. Current therapy approaches for obesity-induced non-communicable diseases persist in prioritizing symptom remission while frequently overlooking the criticality of targeting and alleviating inflammation at its source. Accordingly, this review highlights the importance of the microenvironment of obese white adipose tissue and the promising potential of employing immunotherapy to target it as an effective therapeutic approach for non-communicable diseases induced by obesity. Additionally, this review discusses the challenges and offers perspective about the immunotherapy targeting the microenvironment of obese white adipose tissue.
Collapse
Affiliation(s)
- Lia Priscilla
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Chaerim Yoo
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seonmi Jang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Sewon Park
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Gayoung Lim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Taekyun Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Nano Science and Technology (INST) & Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, 04763, Republic of Korea
- Elixir Pharmatech Inc., Seoul, 07463, Republic of Korea
| |
Collapse
|
29
|
Shimizu I. Exploration of New Therapies for Heart Failure Targeting Age-Related Mechanisms. Circ J 2024; 88:626-630. [PMID: 37438143 DOI: 10.1253/circj.cj-23-0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 07/14/2023]
Abstract
Evidence indicates a role of cellular senescence and systemic insulin resistance (hyperinsulinemia) in the pathogenesis of age-related cardiovascular-metabolic disorders, including heart failure, atherosclerotic diseases, obesity, and diabetes. "Metabolic remodeling" is one of the keywords for aging research, and studies with brown adipose tissue have shown that maintaining the homeostasis of this organ is crucial to suppressing the progression of pathologies in obesity and heart failure. The mechanisms contributing to the synchronization of aging (sync-aging) are mysterious and interesting. "Senometabolite" or "senoprotein" are defined as circulating molecules that have causal roles in sync-aging, which requires the establishment of new concepts: age-related fibrotic disorders (A-FiDs), and senometabolite-related disorders (SRDs). Globally, researchers are active in comprehensive and conclusive studies targeting age-related circulating molecules. Recently, the senolytic approach opened a new avenue for aging research. Senolysis, mediated through a genetic/pharmacologic/vaccination approach, reversed aging and pathologies in age-related diseases. Suppression of prosenescent molecules (senocules) and senolysis, the specific depletion of senescent cells, will become next-generation therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| |
Collapse
|
30
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: When immune cells help to re-build tissues. FEBS J 2024; 291:1597-1614. [PMID: 36440547 PMCID: PMC10225019 DOI: 10.1111/febs.16693] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is an essential immune response critical for responding to infection, injury and maintenance of tissue homeostasis. Upon injury, regenerative inflammation promotes tissue repair by a timed and coordinated infiltration of diverse cell types and the secretion of growth factors, cytokines and lipids mediators. Remarkably, throughout evolution as well as mammalian development, this type of physiological inflammation is highly associated with immunosuppression. For instance, regenerative inflammation is the consequence of an in situ macrophage polarization resulting in a transition from pro-inflammatory to anti-inflammatory/pro-regenerative response. Immune cells are the first responders upon injury, infiltrating the damaged tissue and initiating a pro-inflammatory response depleting cell debris and necrotic cells. After phagocytosis, macrophages undergo multiple coordinated metabolic and transcriptional changes allowing the transition and dictating the initiation of the regenerative phase. Differences between a highly efficient, complete ad integrum tissue repair, such as, acute skeletal muscle injury, and insufficient regenerative inflammation, as the one developing in Duchenne Muscular Dystrophy (DMD), highlight the importance of a coordinated response orchestrated by immune cells. During regenerative inflammation, these cells interact with others and alter the niche, affecting the character of inflammation itself and, therefore, the progression of tissue repair. Comparing acute muscle injury and chronic inflammation in DMD, we review how the same cells and molecules in different numbers, concentration and timing contribute to very different outcomes. Thus, it is important to understand and identify the distinct functions and secreted molecules of macrophages, and potentially other immune cells, during tissue repair, and the contributors to the macrophage switch leveraging this knowledge in treating diseases.
Collapse
Affiliation(s)
- Noemí Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology, Faculty of Medicine, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
| | - Sergio Alonso-Alonso
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| |
Collapse
|
32
|
Rosendo-Silva D, Gomes PB, Rodrigues T, Viana S, da Costa AN, Scherer PE, Reis F, Pereira F, Seiça R, Matafome P. Clinical and molecular profiling of human visceral adipose tissue reveals impairment of vascular architecture and remodeling as an early hallmark of dysfunction. Metabolism 2024; 153:155788. [PMID: 38219974 DOI: 10.1016/j.metabol.2024.155788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/03/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Adipose tissue dysfunction is more related to insulin resistance than body mass index itself and an alteration in adipose tissue function is thought to underlie the shift from metabolically healthy to unhealthy obesity. Herein, we performed a clustering analysis that revealed distinct visceral adipose tissue gene expression patterns in patients with obesity at distinct stages of metabolic dysregulation. We have built a cross-sectional cohort that aims at reflecting the evolution of the metabolic sequelae of obesity with the main objective to map the sequential events that play a role in adipose tissue dysfunction from the metabolically healthy (insulin-sensitive) state to several incremental degrees of metabolic dysregulation, encompassing insulin resistance establishment, pre-diabetes, and type 2 diabetes. We found that insulin resistance is mainly marked by the downregulation of adipose tissue vasculature remodeling-associated gene expression, suggesting that processes like angiogenesis and adaptative expansion/retraction ability suffer early dysregulation. Prediabetes was characterized by compensatory growth factor-dependent signaling and increased response to hypoxia, while type 2 diabetes was associated with loss of cellular response to insulin and hypoxia and concomitant upregulation of inflammatory markers. Our findings suggest a putative sequence of dysregulation of biological processes that is not linear and has multiple distinct phases across the metabolic dysregulation process, ultimately culminating in the climax of adipose tissue dysfunction in type 2 diabetes. Several studies have addressed the transcriptomic changes in adipose tissue of patients with obesity. However, to the best of our knowledge, this is the first study unraveling the potential molecular mechanisms associated with the multi-step evolution of adipose tissue dysfunction along the metabolic sequelae of obesity.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Pedro Bastos Gomes
- Department of Surgery, Universitary Hospital Center of Coimbra, Portugal
| | - Tiago Rodrigues
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Sofia Viana
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - André Nogueira da Costa
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Translational Medicine, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Flávio Reis
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Francisco Pereira
- Polytechnic University of Coimbra, Coimbra Institute of Engineering, Coimbra, Portugal; Centre for Informatics and Systems of the University of Coimbra (CISUC), University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
Collapse
|
33
|
Wu Y, Ma Y. CCL2-CCR2 signaling axis in obesity and metabolic diseases. J Cell Physiol 2024; 239:e31192. [PMID: 38284280 DOI: 10.1002/jcp.31192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/14/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.
Collapse
Affiliation(s)
- Yue Wu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yanchun Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
34
|
Chaurasiya V, Nidhina Haridas PA, Olkkonen VM. Adipocyte-endothelial cell interplay in adipose tissue physiology. Biochem Pharmacol 2024; 222:116081. [PMID: 38408682 DOI: 10.1016/j.bcp.2024.116081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Adipose tissue (AT) expansion through hyperplasia or hypertrophy requires vascular remodeling that involves angiogenesis. There is quite some evidence that obese white AT (WAT) displays altered vasculature. Some studies suggest that this is associated with hypoxia, which is thought to play a role in inducing inflammatory activation of the excessively expanding WAT. Increasing evidence, based on genetic manipulations or treatments with inhibitory or activator pharmaceuticals, demonstrates that AT angiogenesis is crucial for AT metabolic function, and thereby for whole body metabolism and metabolic health. Despite some contradiction between studies, disturbance of WAT angiogenesis in obesity could be an important factor driving WAT dysfunction and the comorbidities of obesity. Endothelial cells (ECs) contribute to healthy WAT metabolism via transport of fatty acids and other plasma components, secretory signaling molecules, and extracellular vesicles (EVs). This communication is crucial for adipocyte metabolism and underscores the key role that the AT endothelium plays in systemic energy homeostasis and healthy metabolism. Adipocytes communicate towards the neighboring endothelium through several mechanisms. The pro-inflammatory status of hypertrophic adipocytes in obesity is reflected in ECs activation, which promotes chronic inflammation. On the other hand, adiponectin secreted by the adipocytes is important for healthy endothelial function, and adipocytes also secrete other pro- or anti-angiogenic effector molecules and a wealth of EVs - however, their detailed roles in signaling towards the endothelium are yet poorly understood. To conclude, targeting AT angiogenesis and promoting the healthy communication between adipocytes and ECs represent potentially promising strategies to treat obesity and its comorbidities.
Collapse
Affiliation(s)
- Vaishali Chaurasiya
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
35
|
Yao T, Wei D, Tian X, Zhao L, Wan Q, Zhang X, Cai J, Li S, Diao B, Feng S, Shan B, Shao M, Wu Y. PDGFRβ + cell HIF2α is dispensable for white adipose tissue metabolic remodeling and hepatic lipid accumulation in obese mice. Lipids Health Dis 2024; 23:81. [PMID: 38509584 PMCID: PMC10953078 DOI: 10.1186/s12944-024-02069-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Obesity is associated with extensive white adipose tissue (WAT) expansion and remodeling. Healthy WAT expansion contributes to the maintenance of energy balance in the liver, thereby ameliorating obesity-related hepatic steatosis. Tissue-resident mesenchymal stromal cell populations, including PDGFRβ + perivascular cells, are increasingly recognized pivotal as determinants of the manner in which WAT expands. However, the full array of regulatory factors controlling WAT stromal cell functions remains to be fully elucidated. Hypoxia-inducible factors (HIFs) are critical regulators in WAT stromal cell populations such as adipocyte precursor cells (APCs). It is revealed that HIF1α activation within PDGFRβ + stromal cells results in the suppression of de novo adipogenesis and the promotion of a pro-fibrogenic cellular program in obese animals. However, the role of HIF2α in PDGFRβ + cells remains undetermined in vivo. METHODS New genetic models were employed in which HIF1α (encoded by the Hif1a gene) and HIF2α (encoded by the Epas1 gene) are selectively inactivated in PDGFRβ + cells in an inducible manner using tamoxifen (TAM). With these models, both in vitro and in vivo functional analysis of PDGFRβ + cells lacking HIF proteins were performed. Additionally, comprehensive metabolic phenotyping in diet-induced mouse models were performed to investigate the roles of PDGFRβ + cell HIF proteins in WAT remodeling, liver energy balance and systemic metabolism. RESULTS Unlike HIF1α inactivation, the new findings in this study suggest that inducible ablation of HIF2α in PDGFRβ + cells does not cause apparent effects on WAT expansion induced by obesogenic diet. The adipogenic ability of PDGFRβ + APCs is not significantly altered by genetic HIF2α ablation. Moreover, no difference of key parameters associated with healthy WAT remodeling such as improvements of WAT insulin sensitivity, reduction in metabolic inflammation, as well as changes in liver fat accumulation or systemic glucose metabolism, is detected in PDGFRβ + cell Epas1-deficient mice. CONCLUSION The new findings in this study support that, in contrast to HIF1α, PDGFRβ + cell HIF2α appears dispensable for WAT metabolic remodeling and the resulting effects on liver metabolic homeostasis in diet-induced obesity, underscoring the isoform-specific roles of HIFα proteins in the regulation of adipose tissue biology.
Collapse
Affiliation(s)
- Tao Yao
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Danni Wei
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Tian
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Lin Zhao
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Qiangyou Wan
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoli Zhang
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Juan Cai
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Siqi Li
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bowen Diao
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Suihan Feng
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Bo Shan
- Cancer Center, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Mengle Shao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
| | - Ying Wu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
36
|
Zhou XT, Zhu AQ, Li XM, Sun LY, Yan JG, Luo N, Chen SS, Huang Z, Mao XL, Li KP. Mulberry and Hippophae-based solid beverage promotes weight loss in rats by antagonizing white adipose tissue PPARγ and FGFR1 signaling. Front Endocrinol (Lausanne) 2024; 15:1344262. [PMID: 38559696 PMCID: PMC10978776 DOI: 10.3389/fendo.2024.1344262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Obesity, a multifactorial disease with many complications, has become a global epidemic. Weight management, including dietary supplementation, has been confirmed to provide relevant health benefits. However, experimental evidence and mechanistic elucidation of dietary supplements in this regard are limited. Here, the weight loss efficacy of MHP, a commercial solid beverage consisting of mulberry leaf aqueous extract and Hippophae protein peptides, was evaluated in a high-fat high-fructose (HFF) diet-induced rat model of obesity. Body component analysis and histopathologic examination confirmed that MHP was effective to facilitate weight loss and adiposity decrease. Pathway enrichment analysis with differential metabolites generated by serum metabolomic profiling suggests that PPAR signal pathway was significantly altered when the rats were challenged by HFF diet but it was rectified after MHP intervention. RNA-Seq based transcriptome data also indicates that MHP intervention rectified the alterations of white adipose tissue mRNA expressions in HFF-induced obese rats. Integrated omics reveals that the efficacy of MHP against obesogenic adipogenesis was potentially associated with its regulation of PPARγ and FGFR1 signaling pathway. Collectively, our findings suggest that MHP could improve obesity, providing an insight into the use of MHP in body weight management.
Collapse
Affiliation(s)
- Xiao-Ting Zhou
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - An-Qi Zhu
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao-Min Li
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Ling-Yue Sun
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jian-Gang Yan
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Nin Luo
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shi-Sheng Chen
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Zebo Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xin-Liang Mao
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Kun-Ping Li
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
37
|
Conte C, Cipponeri E, Roden M. Diabetes Mellitus, Energy Metabolism, and COVID-19. Endocr Rev 2024; 45:281-308. [PMID: 37934800 PMCID: PMC10911957 DOI: 10.1210/endrev/bnad032] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/10/2023] [Revised: 08/30/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Obesity, diabetes mellitus (mostly type 2), and COVID-19 show mutual interactions because they are not only risk factors for both acute and chronic COVID-19 manifestations, but also because COVID-19 alters energy metabolism. Such metabolic alterations can lead to dysglycemia and long-lasting effects. Thus, the COVID-19 pandemic has the potential for a further rise of the diabetes pandemic. This review outlines how preexisting metabolic alterations spanning from excess visceral adipose tissue to hyperglycemia and overt diabetes may exacerbate COVID-19 severity. We also summarize the different effects of SARS-CoV-2 infection on the key organs and tissues orchestrating energy metabolism, including adipose tissue, liver, skeletal muscle, and pancreas. Last, we provide an integrative view of the metabolic derangements that occur during COVID-19. Altogether, this review allows for better understanding of the metabolic derangements occurring when a fire starts from a small flame, and thereby help reducing the impact of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome 00166, Italy
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Elisa Cipponeri
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg 85764, Germany
| |
Collapse
|
38
|
Nicholas DA, Mbongue JC, Garcia-Pérez D, Sorensen D, Ferguson Bennit H, De Leon M, Langridge WHR. Exploring the Interplay between Fatty Acids, Inflammation, and Type 2 Diabetes. IMMUNO 2024; 4:91-107. [PMID: 39606781 PMCID: PMC11600342 DOI: 10.3390/immuno4010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2024] Open
Abstract
Around 285 million people worldwide currently have type 2 diabetes and it is projected that this number will be surpassed by 2030. Therefore, it is of the utmost importance to enhance our comprehension of the disease's development. The regulation of diet, obesity, and inflammation in type 2 diabetes is believed to play a crucial role in enhancing insulin sensitivity and reducing the risk of onset diabetes. Obesity leads to an increase in visceral adipose tissue, which is a prominent site of inflammation in type 2 diabetes. Dyslipidemia, on the other hand, plays a significant role in attracting activated immune cells such as macrophages, dendritic cells, T cells, NK cells, and B cells to visceral adipose tissue. These immune cells are a primary source of pro-inflammatory cytokines that are believed to promote insulin resistance. This review delves into the influence of elevated dietary free saturated fatty acids and examines the cellular and molecular factors associated with insulin resistance in the initiation of inflammation induced by obesity. Furthermore, it explores novel concepts related to diet-induced inflammation and its relationship with type 2 diabetes.
Collapse
Affiliation(s)
- Dequina A. Nicholas
- School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Jacques C. Mbongue
- Department of Biological Sciences, School of Arts and Sciences, Oakwood University, Huntsville, AL 35896, USA
| | - Darysbel Garcia-Pérez
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
- Division of Molecular Genetics and Microbiology, School of Medicine Alumni Hall, Loma Linda University, Rm 102, 11021 Campus Street, Loma Linda, CA 92350, USA
| | - Dane Sorensen
- Center for Perinatal Biology, Division of Physiology, Loma Linda School of Medicine, Rm A572, 11234 Anderson Street, Loma Linda, CA 92350, USA
| | - Heather Ferguson Bennit
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| | - Marino De Leon
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| | - William H. R. Langridge
- Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 11085, USA
| |
Collapse
|
39
|
Bi J, Zhou W, Tang Z. Pathogenesis of diabetic complications: Exploring hypoxic niche formation and HIF-1α activation. Biomed Pharmacother 2024; 172:116202. [PMID: 38330707 DOI: 10.1016/j.biopha.2024.116202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/11/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Hypoxia is a common feature of diabetic tissues, which highly correlates to the progression of diabetes. The formation of hypoxic context is induced by disrupted oxygen homeostasis that is predominantly driven by vascular remodeling in diabetes. While different types of vascular impairments have been reported, the specific features and underlying mechanisms are yet to be fully understood. Under hypoxic condition, cells upregulate hypoxia-inducible factor-1α (HIF-1α), an oxygen sensor that coordinates oxygen concentration and cell metabolism under hypoxic conditions. However, diabetic context exploits this machinery for pathogenic functions. Although HIF-1α protects cells from diabetic insult in multiple tissues, it also jeopardizes cell function in the retina. To gain a deeper understanding of hypoxia in diabetic complications, we focus on the formation of tissue hypoxia and the outcomes of HIF-1α dysregulation under diabetic context. Hopefully, this review can provide a better understanding on hypoxia biology in diabetes.
Collapse
Affiliation(s)
- Jingjing Bi
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education,Southwest Medical University, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Wenhao Zhou
- Yucebio Technology Co., Ltd., Shenzhen, China
| | - Zonghao Tang
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education,Southwest Medical University, Ministry of Education, Southwest Medical University, Luzhou, China; Baylor College of Medicine, Department of Molecular and Cellular Biology, Houston, TX, USA.
| |
Collapse
|
40
|
Chandrasekaran P, Weiskirchen R. The Role of Obesity in Type 2 Diabetes Mellitus-An Overview. Int J Mol Sci 2024; 25:1882. [PMID: 38339160 PMCID: PMC10855901 DOI: 10.3390/ijms25031882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity or excessive weight gain is identified as the most important and significant risk factor in the development and progression of type 2 diabetes mellitus (DM) in all age groups. It has reached pandemic dimensions, making the treatment of obesity crucial in the prevention and management of type 2 DM worldwide. Multiple clinical studies have demonstrated that moderate and sustained weight loss can improve blood glucose levels, insulin action and reduce the need for diabetic medications. A combined approach of diet, exercise and lifestyle modifications can successfully reduce obesity and subsequently ameliorate the ill effects and deadly complications of DM. This approach also helps largely in the prevention, control and remission of DM. Obesity and DM are chronic diseases that are increasing globally, requiring new approaches to manage and prevent diabetes in obese individuals. Therefore, it is essential to understand the mechanistic link between the two and design a comprehensive approach to increase life expectancy and improve the quality of life in patients with type 2 DM and obesity. This literature review provides explicit information on the clinical definitions of obesity and type 2 DM, the incidence and prevalence of type 2 DM in obese individuals, the indispensable role of obesity in the pathophysiology of type 2 DM and their mechanistic link. It also discusses clinical studies and outlines the recent management approaches for the treatment of these associated conditions. Additionally, in vivo studies on obesity and type 2 DM are discussed here as they pave the way for more rigorous development of therapeutic approaches.
Collapse
Affiliation(s)
- Preethi Chandrasekaran
- UT Southwestern Medical Center Dallas, 5323 Harry Hines Blvd. ND10.504, Dallas, TX 75390-9014, USA
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH), University Hospital Aachen, D-52074 Aachen, Germany
| |
Collapse
|
41
|
Bilson J, Oquendo CJ, Read J, Scorletti E, Afolabi PR, Lord J, Bindels LB, Targher G, Mahajan S, Baralle D, Calder PC, Byrne CD, Sethi JK. Markers of adipose tissue fibrogenesis associate with clinically significant liver fibrosis and are unchanged by synbiotic treatment in patients with NAFLD. Metabolism 2024; 151:155759. [PMID: 38101770 DOI: 10.1016/j.metabol.2023.155759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/27/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND AND AIMS Subcutaneous adipose tissue (SAT) dysfunction contributes to NAFLD pathogenesis and may be influenced by the gut microbiota. Whether transcript profiles of SAT are associated with liver fibrosis and are influenced by synbiotic treatment (that changes the gut microbiome) is unknown. We investigated: (a) whether the presence of clinically significant, ≥F2 liver fibrosis associated with adipose tissue (AT) dysfunction, differential gene expression in SAT, and/or a marker of tissue fibrosis (Composite collagen gene expression (CCGE)); and (b) whether synbiotic treatment modified markers of AT dysfunction and the SAT transcriptome. METHODS Sixty-two patients with NAFLD (60 % men) were studied before and after 12 months of treatment with synbiotic or placebo and provided SAT samples. Vibration-controlled transient elastography (VCTE)-validated thresholds were used to assess liver fibrosis. RNA-sequencing and histological analysis of SAT were performed to determine differential gene expression, CCGE and the presence of collagen fibres. Regression modelling and receiver operator characteristic curve analysis were used to test associations with, and risk prediction for, ≥F2 liver fibrosis. RESULTS Patients with ≥F2 liver fibrosis (n = 24) had altered markers of AT dysfunction and a SAT gene expression signature characterised by enrichment of inflammatory and extracellular matrix-associated genes, compared to those with CONCLUSION A differential gene expression signature in SAT associates with ≥F2 liver fibrosis is explained by a measure of systemic insulin resistance and is not changed by synbiotic treatment. SAT CCGE values are a good predictor of ≥F2 liver fibrosis in NAFLD.
Collapse
Affiliation(s)
- Josh Bilson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
| | - Carolina J Oquendo
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - James Read
- School of Chemistry, Faculty of Engineering and Physical sciences, University of Southampton, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Eleonora Scorletti
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK; Division of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul R Afolabi
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
| | - Jenny Lord
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UC Louvain, Université Catholique de Louvain, Brussels, Belgium; Welbio department, WEL Research Institute, Wavre, Belgium
| | - Giovanni Targher
- Department of Medicine, University of Verona, Italy; Metabolic Diseases Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Sumeet Mahajan
- School of Chemistry, Faculty of Engineering and Physical sciences, University of Southampton, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Diana Baralle
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christopher D Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK.
| | - Jaswinder K Sethi
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
42
|
Hai Y, Ren K, Zhang Y, Yang L, Cao H, Yuan X, Su L, Li H, Feng X, Liu D. HIF-1α serves as a co-linker between AD and T2DM. Biomed Pharmacother 2024; 171:116158. [PMID: 38242039 DOI: 10.1016/j.biopha.2024.116158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/24/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Alzheimer's disease (AD)-related brain deterioration is linked to the type 2 diabetes mellitus (T2DM) features hyperglycemia, hyperinsulinemia, and insulin resistance. Hypoxia as a common risk factor for both AD and T2DM. Hypoxia-inducible factor-1 alpha (HIF-1α) acts as the main regulator of the hypoxia response and may be a key target in the comorbidity of AD and T2DM. HIF-1α expression is closely related to hyperglycemia, insulin resistance, and inflammation. Tissue oxygen consumption disrupts HIF-1α homeostasis, leading to increased reactive oxygen species levels and the inhibition of insulin receptor pathway activity, causing neuroinflammation, insulin resistance, abnormal Aβ deposition, and tau hyperphosphorylation. HIF-1α activation also leads to the deposition of Aβ by promoting the abnormal shearing of amyloid precursor protein and inhibiting the degradation of Aβ, and it promotes tau hyperphosphorylation by activating oxidative stress and the activation of astrocytes, which further exasperates AD. Therefore, we believe that HIF-α has great potential as a target for the treatment of AD. Importantly, the intracellular homeostasis of HIF-1α is a more crucial factor than its expression level.
Collapse
Affiliation(s)
- Yang Hai
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Key Laboratory of Dunhuang Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China.
| | - Ke Ren
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Yarong Zhang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Lili Yang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Haoshi Cao
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Xianxia Yuan
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Linling Su
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Hailong Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Xiaoli Feng
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Key Laboratory of Dunhuang Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Dongling Liu
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Northwest Collaborative Innovation Center for Traditional Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Gansu Pharmaceutical Industry Innovation Research Institute, Lanzhou 730000, Gansu Province, PR China.
| |
Collapse
|
43
|
Balan AI, Halațiu VB, Scridon A. Oxidative Stress, Inflammation, and Mitochondrial Dysfunction: A Link between Obesity and Atrial Fibrillation. Antioxidants (Basel) 2024; 13:117. [PMID: 38247541 PMCID: PMC10812976 DOI: 10.3390/antiox13010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
The adipose tissue has long been thought to represent a passive source of triglycerides and fatty acids. However, extensive data have demonstrated that the adipose tissue is also a major endocrine organ that directly or indirectly affects the physiological functions of almost all cell types. Obesity is recognized as a risk factor for multiple systemic conditions, including metabolic syndrome, type 2 diabetes mellitus, sleep apnea, cardiovascular disorders, and many others. Obesity-related changes in the adipose tissue induce functional and structural changes in cardiac myocytes, promoting a wide range of cardiovascular disorders, including atrial fibrillation (AF). Due to the wealth of epidemiologic data linking AF to obesity, the mechanisms underlying AF occurrence in obese patients are an area of rich ongoing investigation. However, progress has been somewhat slowed by the complex phenotypes of both obesity and AF. The triad inflammation, oxidative stress, and mitochondrial dysfunction are critical for AF pathogenesis in the setting of obesity via multiple structural and functional proarrhythmic changes at the level of the atria. The aim of this paper is to provide a comprehensive view of the close relationship between obesity-induced oxidative stress, inflammation, and mitochondrial dysfunction and the pathogenesis of AF. The clinical implications of these mechanistic insights are also discussed.
Collapse
Affiliation(s)
- Alkora Ioana Balan
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Vasile Bogdan Halațiu
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Alina Scridon
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| |
Collapse
|
44
|
Engin A. Adipose Tissue Hypoxia in Obesity: Clinical Reappraisal of Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:329-356. [PMID: 39287857 DOI: 10.1007/978-3-031-63657-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 09/19/2024]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases β cell death. Moreover, deletion of HIF-1α worsens β cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
45
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
46
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
47
|
Engin AB. Message Transmission Between Adipocyte and Macrophage in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:273-295. [PMID: 39287855 DOI: 10.1007/978-3-031-63657-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 09/19/2024]
Abstract
Obesity is characterized by the chronic low-grade activation of the innate immune system. In this respect, macrophage-elicited metabolic inflammation and adipocyte-macrophage interaction have primary importance in obesity. Large quantity of macrophages is accumulated by different mechanisms in obese adipose tissue. Hypertrophic adipocyte-derived chemotactic monocyte chemoattractant protein-1 (MCP-1)/C-C chemokine receptor 2 (CCR2) pathway promotes more macrophage accumulation into the obese adipose tissue. However, obesity-induced changes in adipose tissue macrophage density are mainly dependent on increases in the triple-positive cluster of differentiation (CD)11b+ F4/80+ CD11c+ adipose tissue macrophage subpopulation. As epigenetic regulators, microRNAs (miRNAs) are one of the most important mediators of obesity. miRNAs are expressed by adipocytes as well as macrophages and regulate inflammation with the expression of target genes. A paracrine loop involving free fatty acids and tumor necrosis factor-alpha (TNF-α) between adipocytes and macrophages establishes a vicious cycle that aggravates inflammatory changes in the adipose tissue. Adipocyte-specific caspase-1 and production of interleukin-1beta (IL-1β) by macrophages; both adipocyte and macrophage induction by toll-like receptor-4 (TLR4) through nuclear factor-kappaB (NF-κB) activation; free fatty acid-induced and TLR-mediated activation of c-Jun N-terminal kinase (JNK)-related pro-inflammatory pathways in CD11c+ immune cells; are effective in mutual message transmission between adipocyte and macrophage and in the development of adipose tissue inflammation. Thus, the metabolic status of adipocytes and their released exosomes are important determinants of macrophage inflammatory output. However, old adipocytes are removed by macrophages through trogocytosis or sending an "eat me" signal. As a single miRNA can be able to regulate a variety of target genes and signaling pathways, reciprocal transfer of miRNAs between adipocytes and macrophages via miRNA-loaded exosomes reorganizes the different stages of obesity. Changes in the expression of circulating miRNAs because of obesity progression or anti-obesity treatment indicate that miRNAs could be used as potential biomarkers. Therefore, it is believed that targeting macrophage-associated miRNAs with anti-obesity miRNA-loaded nano-carriers may be successful in the attenuation of both obesity and adipose tissue inflammation in clinical practice. Moreover, miRNA-containing exosomes and transferable mitochondria between the adipocyte and macrophage are investigated as new therapeutic targets for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
48
|
Engin A. Bariatric Surgery in Obesity: Metabolic Quality Analysis and Comparison of Surgical Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:697-726. [PMID: 39287870 DOI: 10.1007/978-3-031-63657-8_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 09/19/2024]
Abstract
Obesity is a constantly growing health problem which reduces quality of life and life expectancy. Bariatric surgery (BS) for obesity is considered when all other conservative treatment modalities have failed. Comparison of the multidisciplinary programs with BS regarding to the weight loss showed that substantial and durable weight reduction have been achieved only with bariatric surgical treatments. Although laparoscopic sleeve gastrectomy is the most popular BS, it has high long-term failure rates, and it is claimed that one of every three patients will undergo another bariatric procedure within a 10-year period. Although BS provides weight loss and improvement of metabolic comorbidities, in long-term follow-up, weight gain is observed in half of the patients, while decrease in bone mass and nutritional deficiencies occur in up to 90%. Moreover, despite significant weight loss, several psychological aspects of patients are worsened in comparison to preoperative levels. Nearly one-fifth of postoperative patients with "Loss-of-eating control" meet food addiction criteria. Therefore, the benefits of weight loss following bariatric procedures alone are still debated in terms of the proinflammatory and metabolic profile of obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
49
|
Kudo T, Zhao ML, Jeknić S, Kovary KM, LaGory EL, Covert MW, Teruel MN. Context-dependent regulation of lipid accumulation in adipocytes by a HIF1α-PPARγ feedback network. Cell Syst 2023; 14:1074-1086.e7. [PMID: 37995680 PMCID: PMC11251692 DOI: 10.1016/j.cels.2023.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/04/2022] [Revised: 12/03/2022] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Hypoxia-induced upregulation of HIF1α triggers adipose tissue dysfunction and insulin resistance in obese patients. HIF1α closely interacts with PPARγ, the master regulator of adipocyte differentiation and lipid accumulation, but there are conflicting results regarding how this interaction controls the excessive lipid accumulation that drives adipocyte dysfunction. To directly address these conflicts, we established a differentiation system that recapitulated prior seemingly opposing observations made across different experimental settings. Using single-cell imaging and coarse-grained mathematical modeling, we show how HIF1α can both promote and repress lipid accumulation during adipogenesis. Our model predicted and our experiments confirmed that the opposing roles of HIF1α are isolated from each other by the positive-feedback-mediated upregulation of PPARγ that drives adipocyte differentiation. Finally, we identify three factors: strength of the differentiation cue, timing of hypoxic perturbation, and strength of HIF1α expression changes that, when considered together, provide an explanation for many of the previous conflicting reports.
Collapse
Affiliation(s)
- Takamasa Kudo
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Michael L Zhao
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Stevan Jeknić
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kyle M Kovary
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Edward L LaGory
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Markus W Covert
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Mary N Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Biochemistry and the Drukier Institute of Children's Health, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
50
|
Chaurasiya V, Pham DD, Harju J, Juuti A, Penttilä A, Emmagouni SKG, Nguyen VD, Zhang B, Perttunen S, Keskitalo S, Zhou Y, Pietiläinen KH, Haridas PAN, Olkkonen VM. Human visceral adipose tissue microvascular endothelial cell isolation and establishment of co-culture with white adipocytes to analyze cell-cell communication. Exp Cell Res 2023; 433:113819. [PMID: 37852349 DOI: 10.1016/j.yexcr.2023.113819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023]
Abstract
Communication between adipocytes and endothelial cells (EC) is suggested to play an important role in the metabolic function of white adipose tissue. In order to generate tools to investigate in detail the physiology and communication of EC and adipocytes, a method for isolation of adipose microvascular EC from visceral adipose tissue (VAT) biopsies of subjects with obesity was developed. Moreover, mature white adipocytes were isolated from the VAT biopsies by a method adapted from a previously published Membrane aggregate adipocytes culture (MAAC) protocol. The identity and functionality of the cultivated and isolated adipose microvascular EC (AMvEC) was validated by imaging their morphology, analyses of mRNA expression, fluorescence activated cell sorting (FACS), immunostaining, low-density lipoprotein (LDL) uptake, and in vitro angiogenesis assays. Finally, we established a new trans filter co-culture system (membrane aggregate adipocyte and endothelial co-culture, MAAECC) for the analysis of communication between the two cell types. EC-adipocyte communication in this system was validated by omics analyses, revealing several altered proteins belonging to pathways such as metabolism, intracellular transport and signal transduction in adipocytes co-cultured with AMvEC. In reverse experiments, induction of several pathways including endothelial development and functions was found in AMvEC co-cultured with adipocytes. In conclusion, we developed a robust method to isolate EC from small quantities of human VAT. Furthermore, the MAAECC system established during the study enables one to study the communication between primary white adipocytes and EC or vice-versa and could also be employed for drug screening.
Collapse
Affiliation(s)
- Vaishali Chaurasiya
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland; Doctoral Programme in Biomedicine, University of Helsinki, Finland.
| | - Dan Duc Pham
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Jukka Harju
- Department of Gastrointestinal Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anne Juuti
- Department of Gastrointestinal Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anne Penttilä
- Department of Gastrointestinal Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Van Dien Nguyen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Birong Zhang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sanni Perttunen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Salla Keskitalo
- Molecular Systems Biology Research Group & Proteomics Unit, HiLIFE Helsinki Institute of Life Science, Institute of Biotechnology, University of Helsinki, Finland
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|